EP3309168A1 - Procédé pour améliorer l'élimination de virus dans la purification de protéines - Google Patents

Procédé pour améliorer l'élimination de virus dans la purification de protéines Download PDF

Info

Publication number
EP3309168A1
EP3309168A1 EP17195123.9A EP17195123A EP3309168A1 EP 3309168 A1 EP3309168 A1 EP 3309168A1 EP 17195123 A EP17195123 A EP 17195123A EP 3309168 A1 EP3309168 A1 EP 3309168A1
Authority
EP
European Patent Office
Prior art keywords
antibody
protein
antibodies
virus
filtration
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP17195123.9A
Other languages
German (de)
English (en)
Inventor
Amit Mehta
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
F Hoffmann La Roche AG
Original Assignee
F Hoffmann La Roche AG
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=42985499&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=EP3309168(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by F Hoffmann La Roche AG filed Critical F Hoffmann La Roche AG
Publication of EP3309168A1 publication Critical patent/EP3309168A1/fr
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/36Extraction; Separation; Purification by a combination of two or more processes of different types
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/06Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies from serum
    • C07K16/065Purification, fragmentation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/16Extraction; Separation; Purification by chromatography
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/16Extraction; Separation; Purification by chromatography
    • C07K1/18Ion-exchange chromatography
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/34Extraction; Separation; Purification by filtration, ultrafiltration or reverse osmosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/06Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies from serum
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans

Definitions

  • the present invention is from the field of protein purification.
  • the invention concerns methods for increasing the filtration capacity of virus filters, by combined use of endotoxin removal and cation-exchange media in the prefiltration process.
  • Mammalian cell lines have become the primary choice for production of recombinant protein therapeutics due to their capacity for proper protein folding and post translational modification such as glycosylation ( Chu and Robinson Current Opinion in Biotechnology 12:180 - 187, 2001 ).
  • these cell lines are also known to contain retrovirus like particles ( Lieber et al. Science 182:56 - 59, 1973 ; Lubiniecki et al. Dev Biol Stand 70:187 - 191, 1989 ) and possess the risk for potential adventitious virus contamination ( Garnick, Dev Biol Stand. Basel: Karger 93:21 - 29, 1998 ).
  • virus removal is typically performed using chromatography and filtration ( Curtis et al., Biotechnology and Bioengineering 84(2):179 - 186, 2003 ).
  • virus filtration removes viruses by size exclusion and is therefore considered a more robust technique. So far usage of virus filtration during downstream purification of biotherapeutics derived from mammalian cell cultures has been limited to removal of retroviruses (80 - 100 nm diameter) due to lack of high throughput membranes with nominal pore size less than 60 nm.
  • the high selectivity and high throughput with parvovirus filters is achieved by casting a thin retentive membrane layer on a microporous substrate.
  • the thin retentive layer while allows very fine separation of proteins and viruses, it is also susceptible to fouling by impurities in the process feedstream resulting in lower filter capacity and flux.
  • the fouling of the virus filters has been attributed to contaminants such as protein aggregates and denatured protein. Bohonak and Zydney ( Bohonak and Zydney, Journal of Membrane Science 254(1-2):71-79, 2005 ) showed that loss in filter capacity could be due to cake formation or pore blockage. Other recent reports ( Bolton et al., Biotechnol. Appl. Biochem.
  • Bolton et al. (Bolton et al. 2006) performed a thorough study that involved testing of several membranes as prefilters and demonstrated that it was possible to increase capacity of normal flow parvovirus (NFP) membranes by almost an order of magnitude by using ViresolveTM depth filter as a prefilter.
  • Brown et al. Brown et al. ( Brown et al. 2008, Use of Charged Membranes to Identify Soluble Protein Foulants in order to Facilitate Parvovirus Filtration.
  • the present invention is based, at least in part, on the experimental finding that fouling of parvovirus filters could be due to impurities other than those mentioned in the literature and more comprehensive prefiltration solutions are required to improve the virus filtration capacity. Accordingly, the present invention provides a novel prefiltration solution that performs significantly better than the best prefiltration approach mentioned in the literature (cation-exchange membrane adsorbers).
  • the invention concerns a method of improving the filtration capacity of a virus filter during protein purification, comprising subjecting a composition comprising a protein to be purified to a cation exchange step and an endotoxin removal step, in either order, prior to passing through said virus filter.
  • the pore size of the virus filter is between about 15 and about 100 nm in diameter.
  • the pore size of the virus filter is between about 15 and about 30 nm in diameter.
  • the pore size of the virus filter is about 20 nm.
  • the virus to be removed is a parvovirus.
  • the diameter of the parvovirus is between about 18 and about 26 nm.
  • the protein is an antibody or an antibody fragment, such as an antibody produced by recombinant DNA techniques, or a fragment thereof.
  • the antibody is a therapeutic antibody.
  • the recombinant antibody or antibody fragment is produced in a mammalian host cell, such as, for example, a Chinese Hamster Ovary (CHO) cell.
  • a mammalian host cell such as, for example, a Chinese Hamster Ovary (CHO) cell.
  • composition comprising the protein to be purified is first subjected to a cation exchange step followed by an endotoxin removal step, prior to virus filtration.
  • composition comprising the protein to be purified is first subjected to an endotoxin removal step followed by a cation exchange step, prior to virus filtration.
  • composition comprising the protein to be purified is subjected to a cation exchange step and endotoxin removal step simultaneously, prior to virus filtration, by keeping the two media together in a single module.
  • the endotoxin removal step is directly followed by virus filtration.
  • the cation exchange step is directly followed by virus filtration.
  • virus filtration is performed at a pH between about 4 and about 10.
  • the protein concentration in the composition to be purified is about 1 - 40 g/L.
  • the antibody to be purified is to one or more antigens selected from the group consisting of HER1 (EGFR), HER2, HER3, HER4, VEGF, CD20, CD22, CD11a, CD11b, CD11c, CD18, an ICAM, VLA-4, VCAM, IL-17A and/or F, IgE, DR5, CD40, Apo2L/TRAIL, EGFL7, NRP1, mitogen activated protein kinase (MAPK), and Factor D.
  • HER1 EGFR
  • HER1 EGFR
  • the antibody is selected from the group consisting of anti-estrogen receptor antibody, anti-progesterone receptor antibody, anti-p53 antibody, anti-cathepsin D antibody, anti-Bcl-2 antibody, anti-E-cadherin antibody, anti-CA125 antibody, anti-CA15-3 antibody, anti-CA19-9 antibody, anti-c-erbB-2 antibody, anti-P-glycoprotein antibody, anti-CEA antibody, anti-retinoblastoma protein antibody, anti-ras oncoprotein antibody, anti-Lewis X antibody, anti-Ki-67 antibody, anti-PCNA antibody, anti-CD3 antibody, anti-CD4 antibody, anti-CD5 antibody, anti-CD7 antibody, anti-CD8 antibody, anti-CD9/p24 antibody, anti-CD10 antibody, anti-CD11c antibody, anti-CD13 antibody, anti-CD14 antibody, anti-CD15 antibody, anti-CD19 antibody, anti-CD23 antibody, anti-CD30 antibody, anti-CD31 antibody, anti-CD
  • protein is meant a sequence of amino acids for which the chain length is sufficient to produce the higher levels of tertiary and/or quaternary structure.
  • proteins are distinguished from “peptides” which are also amino acid - based molecules that do not have such structure.
  • a protein for use herein will have a molecular weight of at least about 15-20 kD, preferably at least about 20 kD.
  • proteins encompassed within the definition herein include mammalian proteins, such as, e.g., CD4, integrins and their subunits, such as beta7, growth hormone, including human growth hormone and bovine growth hormone; growth hormone releasing factor; parathyroid hormone; thyroid stimulating hormone; lipoproteins; ⁇ -1-antitrypsin; insulin A-chain; insulin B-chain; proinsulin; follicle stimulating hormone; calcitonin; luteinizing hormone; glucagon; clotting factors such as factor VIIIC, factor IX, tissue factor, and von Willebrands factor; anti-clotting factors such as Protein C; atrial natriuretic factor; lung surfactant; a plasminogen activator, such as urokinase or tissue-type plasminogen activator (t-PA, e.g ., Activase®, TNKase®, Retevase®); bombazine; thrombin; tumor necrosis factor- ⁇ and
  • therapeutic antibodies and immunoadhesins including, without limitation, antibodies to one or more of the following antigens: HER1 (EGFR), HER2, HER3, HER4, VEGF, CD20, CD22, CD11a, CD11b, CD11c, CD18, an ICAM, VLA-4, VCAM.
  • HER1 EGFR
  • HER2, HER3, HER4, VEGF CD20, CD22, CD11a, CD11b, CD11c, CD18, an ICAM, VLA-4, VCAM.
  • IL-17A and/or F IgE, DR5, CD40, Apo2L/TRAIL, EGFL7, NRP1, mitogen activated protein kinase (MAPK), and Factor D, and fragments thereof.
  • MAPK mitogen activated protein kinase
  • exemplary antibodies include those selected from, and without limitation, anti-estrogen receptor antibody, anti-progesterone receptor antibody, anti-p53 antibody, anti-cathepsin D antibody, anti-Bcl-2 antibody, anti-E-cadherin antibody, anti-CA125 antibody, anti-CA15-3 antibody, anti-CA19-9 antibody, anti-c-erbB-2 antibody, anti-P-glycoprotein antibody, anti-CEA antibody, anti-retinoblastoma protein antibody, anti-ras oncoprotein antibody, anti-Lewis X antibody, anti-Ki-67 antibody, anti-PCNA antibody, anti-CD3 antibody, anti-CD4 antibody, anti-CD5 antibody, anti-CD7 antibody, anti-CD8 antibody, anti-CD9/p24 antibody, anti-CDIO antibody, anti-CD11c antibody, anti-CD13 antibody, anti-CD14 antibody, anti-CD15 antibody, anti-CD19 antibody, anti-CD23 antibody, anti-CD30 antibody, anti-CD31 antibody, anti-CD33 antibody,
  • an "isolated" protein such as antibody
  • Contaminant components of its natural environment are materials which would interfere with diagnostic or therapeutic uses for the protein, such as antibody, and may include enzymes, hormones, and other proteinaceous or nonproteinaceous solutes.
  • the protein such as antibody
  • the protein is preferably essentially pure and desirably essentially homogeneous (i.e. free from contaminating proteins).
  • Essentially pure protein means a composition comprising at least about 90% by weight of the protein, based on total weight of the composition, preferably at least about 95% by weight.
  • Essentially homogeneous protein means a composition comprising at least about 99% by weight of protein, based on total weight of the composition.
  • antibody is used in the broadest sense and specifically covers monoclonal antibodies (including full length antibodies which have an immunoglobulin Fc region), antibody compositions with polyepitopic specificity, bispecific antibodies, diabodies, and single-chain molecules, as well as antibody fragments (e.g ., Fab, F(ab') 2 , and Fv).
  • the basic 4-chain antibody unit is a heterotetrameric glycoprotein composed of two identical light (L) chains and two identical heavy (H) chains.
  • An IgM antibody consists of 5 of the basic heterotetramer unit along with an additional polypeptide called a J chain, and contains 10 antigen binding sites, while IgA antibodies comprise from 2-5 of the basic 4-chain units which can polymerize to form polyvalent assemblages in combination with the J chain.
  • the 4-chain unit is generally about 150,000 daltons.
  • Each L chain is linked to an H chain by one covalent disulfide bond, while the two H chains are linked to each other by one or more disulfide bonds depending on the H chain isotype.
  • Each H and L chain also has regularly spaced intrachain disulfide bridges.
  • Each H chain has at the N-terminus, a variable domain (V H ) followed by three constant domains (C H ) for each of the ⁇ and ⁇ chains and four C H domains for ⁇ and ⁇ isotypes.
  • Each L chain has at the N-terminus, a variable domain (V L ) followed by a constant domain at its other end.
  • the V L is aligned with the V H and the C L is aligned with the first constant domain of the heavy chain (C H 1). Particular amino acid residues are believed to form an interface between the light chain and heavy chain variable domains.
  • the pairing of a V H and V L together forms a single antigen-binding site.
  • immunoglobulins can be assigned to different classes or isotypes. There are five classes of immunoglobulins: IgA, IgD, IgE, IgG and IgM, having heavy chains designated ⁇ , ⁇ , ⁇ , ⁇ and ⁇ , respectively.
  • the ⁇ and ⁇ classes are further divided into subclasses on the basis of relatively minor differences in the CH sequence and function, e.g ., humans express the following subclasses: IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2.
  • variable refers to the fact that certain segments of the variable domains differ extensively in sequence among antibodies.
  • the V domain mediates antigen binding and defines the specificity of a particular antibody for its particular antigen.
  • variability is not evenly distributed across the entire span of the variable domains. Instead, the V regions consist of relatively invariant stretches called framework regions (FRs) of about 15-30 amino acid residues separated by shorter regions of extreme variability called “hypervariable regions” or sometimes “complementarity determining regions” (CDRs) that are each approximately 9-12 amino acid residues in length.
  • FRs framework regions
  • hypervariable regions or sometimes “complementarity determining regions”
  • variable domains of native heavy and light chains each comprise four FRs, largely adopting a ⁇ -sheet configuration, connected by three hypervariable regions, which form loops connecting, and in some cases forming part of, the ⁇ -sheet structure.
  • the hypervariable regions in each chain are held together in close proximity by the FRs and, with the hypervariable regions from the other chain, contribute to the formation of the antigen binding site of antibodies (see Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD (1991 ).
  • the constant domains are not involved directly in binding an antibody to an antigen, but exhibit various effector functions, such as participation of the antibody dependent cellular cytotoxicity (ADCC).
  • ADCC antibody dependent cellular cytotoxicity
  • hypervariable region also known as “complementarity determining regions” or CDRs
  • CDRs complementarity determining regions
  • two residue identification techniques define regions of overlapping, but not identical regions, they can be combined to define a hybrid CDR.
  • the term "monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to conventional (polyclonal) antibody preparations which typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen. In addition to their specificity, the monoclonal antibodies are advantageous in that they are synthesized by the hybridoma culture, uncontaminated by other immunoglobulins.
  • the modifier "monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler et al., Nature, 256: 495 (1975 ), or may be made by recombinant DNA methods (see, e.g., U.S. Patent No. 4,816,567 ).
  • the "monoclonal antibodies” may also be isolated from phage antibody libraries using the techniques described in Clackson et al., Nature, 352:624-628 (1991 ) and Marks et al., J. Mol. Biol., 222:581-597 (1991 ), for example.
  • the monoclonal antibodies herein specifically include "chimeric" antibodies (immunoglobulins) in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is(are) identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity ( U.S. Patent No. 4,816,567 ; Morrison et al., Proc. Natl. Acad. Sci. USA, 81:6851-6855 (1984 )).
  • chimeric antibodies immunoglobulins in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is(are) identical with or homo
  • an “intact” antibody is one which comprises an antigen-binding site as well as a CL and at least the heavy chain domains, C H 1, C H 2 and C H 3.
  • antibody fragment comprises a portion of an intact antibody, preferably the antigen binding and/or the variable region of the intact antibody.
  • antibody fragments include Fab, Fab', F(ab') 2 and Fv fragments; diabodies; linear antibodies (see U.S. Patent 5,641,870 , Example 2; Zapata et al., Protein Eng. 8(10): 1057-1062 [1995 ]); single-chain antibody molecules and multispecific antibodies formed from antibody fragments.
  • Papain digestion of antibodies produced two identical antigen-binding fragments, called “Fab” fragments, and a residual "Fc” fragment, a designation reflecting the ability to crystallize readily.
  • the Fab fragment consists of an entire L chain along with the variable region domain of the H chain (V H ), and the first constant domain of one heavy chain (C H 1).
  • Each Fab fragment is monovalent with respect to antigen binding, i.e., it has a single antigen-binding site.
  • Pepsin treatment of an antibody yields a single large F(ab') 2 fragment which roughly corresponds to two disulfide linked Fab fragments having different antigen-binding activity and is still capable of cross-linking antigen.
  • Fab' fragments differ from Fab fragments by having a few additional residues at the carboxy terminus of the C H 1 domain including one or more cysteines from the antibody hinge region.
  • Fab'-SH is the designation herein for Fab' in which the cysteine residue(s) of the constant domains bear a free thiol group.
  • F(ab') 2 antibody fragments originally were produced as pairs of Fab' fragments which have hinge cysteines between them. Other chemical couplings of antibody fragments are also known.
  • the Fc fragment comprises the carboxy-terminal portions of both H chains held together by disulfides.
  • the effector functions of antibodies are determined by sequences in the Fc region, the region which is also recognized by Fc receptors (FcR) found on certain types of cells.
  • Fv is the minimum antibody fragment which contains a complete antigen-recognition and -binding site. This fragment consists of a dimer of one heavy- and one light-chain variable region domain in tight, non-covalent association. From the folding of these two domains emanate six hypervarible loops (3 loops each from the H and L chain) that contribute the amino acid residues for antigen binding and confer antigen binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three CDRs specific for an antigen) has the ability to recognize and bind antigen, although at a lower affinity than the entire binding site.
  • Single-chain Fv also abbreviated as “sFv” or “scFv” are antibody fragments that comprise the VH and VL antibody domains connected into a single polypeptide chain.
  • the sFv polypeptide further comprises a polypeptide linker between the V H and V L domains which enables the sFv to form the desired structure for antigen binding.
  • diabodies refers to small antibody fragments prepared by constructing sFv fragments (see preceding paragraph) with short linkers (about 5-10) residues) between the V H and V L domains such that inter-chain but not intra-chain pairing of the V domains is achieved, thereby resulting in a bivalent fragment, i.e., a fragment having two antigen-binding sites.
  • Bispecific diabodies are heterodimers of two "crossover" sFv fragments in which the V H and V L domains of the two antibodies are present on different polypeptide chains.
  • Diabodies are described in greater detail in, for example, EP 404,097 ; WO 93/11161 ; Hollinger et al., Proc. Natl. Acad. Sci. USA 90: 6444-6448 (1993 ).
  • An antibody "which binds" a molecular target or an antigen of interest is one capable of binding that antigen with sufficient affinity such that the antibody is useful in targeting a cell expressing the antigen.
  • an antibody that "specifically binds to" or is “specific for” a particular polypeptide or an epitope on a particular polypeptide is one that binds to that particular polypeptide or epitope on a particular polypeptide without substantially binding to any other polypeptide or polypeptide epitope.
  • the extent of binding of the antibody to these other polypeptides or polypeptide epitopes will be less than 10%, as determined by fluorescence activated cell sorting (FACS) analysis or radioimmunoprecipitation (RIA), relative to binding to the target polypeptide or epitope.
  • Humanized forms of non-human (e.g ., murine) antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab', F(ab') 2 or other antigen-binding subsequences of antibodies) of mostly human sequences, which contain minimal sequence derived from non-human immunoglobulin.
  • humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a hypervariable region (also CDR) of the recipient are replaced by residues from a hypervariable region of a non-human species (donor antibody) such as mouse, rat or rabbit having the desired specificity, affinity, and capacity.
  • Fv framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues.
  • "humanized antibodies” as used herein may also comprise residues which are found neither in the recipient antibody nor the donor antibody. These modifications are made to further refine and optimize antibody performance.
  • the humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • Fc immunoglobulin constant region
  • Antibody effector functions refer to those biological activities attributable to the Fc region (a native sequence Fc region or amino acid sequence variant Fc region) of an antibody, and vary with the antibody isotype. Examples of antibody effector functions include: C1q binding and complement dependent cytotoxicity; Fc receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; down regulation of cell surface receptors (e.g., B cell receptors); and B cell activation.
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • FcRs Fc receptors
  • cytotoxic cells e.g., natural killer (NK) cells, neutrophils and macrophages
  • NK cells natural killer cells
  • monocytes express Fc ⁇ RI, Fc ⁇ RII and Fc ⁇ RIII.
  • Fc expression on hematopoietic cells is summarized in Table 3 on page 464 of Ravetch and Kinet, Annu. Rev. Immunol. 9: 457-92 (1991 ).
  • an in vitro ACDD assay such as that described in U.S. Patent No. 5,500,362 or 5,821,337 may be performed.
  • Useful effector cells for such assays include peripheral blood mononuclear cells (PBMC) and natural killer (NK) cells.
  • PBMC peripheral blood mononuclear cells
  • NK natural killer
  • ADCC activity of the molecule of interest may be assessed in vivo, e.g., in an animal model such as that disclosed in Clynes et al., PNAS USA 95:652-656 (1998 ).
  • Fc receptor or “FcR” describes a receptor that binds to the Fc region of an antibody.
  • the preferred FcR is a native sequence human FcR.
  • a preferred FcR is one which binds an IgG antibody (a gamma receptor) and includes receptors of the Fc ⁇ RI, Fc ⁇ RII, and Fc ⁇ RIII subclasses, including allelic variants and alternatively spliced forms of these receptors, Fc ⁇ RII receptors include Fc ⁇ RIIA (an “activating receptor”) and Fc ⁇ RIIB (an “inhibiting receptor”), which have similar amino acid sequences that differ primarily in the cytoplasmic domains thereof.
  • Activating receptor Fc ⁇ RIIA contains an immunoreceptor tyrosine-based activation motif (ITAM) in its cytoplasmic domain.
  • Inhibiting receptor Fc ⁇ RIIB contains an immunoreceptor tyrosine-based inhibition motif (ITIM) in its cytoplasmic domain, (see M. Da ⁇ ron, Annu. Rev. Immunol. 15:203-234 (1997 ).
  • FcRs are reviewed in Ravetch and Kinet, Annu. Rev. Immunol. 9: 457-92 (1991 ); Capel et al., Immunomethods 4: 25-34 (1994 ); and de Haas et al., J. Lab. Clin. Med. 126: 330-41 (1995 ).
  • FcR FcR
  • the term also includes the neonatal receptor, FcRn, which is responsible for the transfer of maternal IgGs to the fetus. Guyer et al., J. Immunol. 117: 587 (1976 ) and Kim et al., J. Immunol. 24: 249 (1994 ).
  • Human effector cells are leukocytes which express one or more FcRs and perform effector functions. Preferably, the cells express at least Fc ⁇ RIII and perform ADCC effector function. Examples of human leukocytes which mediate ADCC include peripheral blood mononuclear cells (PBMC), natural killer (NK) cells, monocytes, cytotoxic T cells and neutrophils, with PBMCs and MNK cells being preferred.
  • PBMC peripheral blood mononuclear cells
  • NK natural killer cells
  • monocytes cytotoxic T cells and neutrophils
  • the effector cells may be isolated from a native source, e.g., blood.
  • “Complement dependent cytotoxicity” of “CDC” refers to the lysis of a target cell in the presence of complement. Activation of the classical complement pathway is initiated by the binding of the first component of the complement system (C1q) to antibodies (of the appropriate subclass) which are bound to their cognate antigen.
  • C1q the first component of the complement system
  • a CDC assay e.g., as described in Gazzano-Santoro et al., J. Immunol. Methods 202: 163 (1996 ), may be performed.
  • conjugates refer to any and all forms of covalent or non-covalent linkage, and include, without limitation, direct genetic or chemical fusion, coupling through a linker or a cross-linking agent, and non-covalent association, for example using a leucine zipper.
  • Antibody conjugates have another entity, such as a cytotoxic compound, drug, composition, compound, radioactive element, or detectable label, attached to an antibody or antibody fragment.
  • Treatment refers to both therapeutic treatment and prophylactic or preventative measures. Those in need of treatment include those already with the disorder as well as those in which the disorder is to be prevented.
  • “Mammal” for purposes of treatment refers to any animal classified as a mammal, including humans, non-human higher primates, domestic and farm animals, and zoo, sports, or pet animals, such as dogs, horses, rabbits, cattle, pigs, hamsters, mice, cats, etc.
  • the mammal is human.
  • a “disorder” is any condition that would benefit from treatment with the protein. This includes chronic and acute disorders or diseases including those pathological conditions which predispose the mammal to the disorder in question.
  • a “therapeutically effective amount” is at least the minimum concentration required to effect a measurable improvement or prevention of a particular disorder.
  • Therapeutically effective amounts of known proteins are well known in the art, while the effective amounts of proteins hereinafter discovered may be determined by standard techniques which are well within the skill of a skilled artisan, such as an ordinary physician.
  • the protein is produced by recombinant DNA techniques, i.e., by culturing cells transformed or transfected with a vector containing nucleic acid encoding the protein, as is well known in art.
  • Preparation of the protein by recombinant means may be accomplished by transfecting or transforming suitable host cells with expression or cloning vectors and cultured in conventional nutrient media modified as appropriate for inducing promoters, selecting transformants, or amplifying the genes encoding the desired sequences.
  • the culture conditions such as media, temperature, pH and the like, can be selected by the skilled artisan without undue experimentation.
  • principles, protocols, and practical techniques for maximizing the productivity of cell cultures can be found in Mammalian Cell Biotechnology: A Practical Approach, M. Butler, Ed. (IRL Press, 1991 ) and Sambrook et al., Molecular Cloning: A Laboratory Manual, New York: Cold Spring Harbor Press .
  • transfection Methods of transfection are known to the ordinarily skilled artisan, and include for example, CaPO 4 and CaCl 2 transfection, electroporation, microinjection, etc. Suitable techniques are also described in Sambrook et al., supra. Additional transfection techniques are described in Shaw et al., Gene 23: 315 (1983 ); WO 89/05859 ; Graham et al., Virology 52: 456-457 (1978 ) and U.S.P. 4,399,216 .
  • the nucleic acid encoding the desired protein may be inserted into a replicable vector for cloning or expression.
  • Suitable vectors are publicly available and may take the form of a plasmid, cosmid, viral particle or phage.
  • the appropriate nucleic acid sequence may be inserted into the vector by a variety of procedures. In general, DNA is inserted into an appropriate restriction endonuclease site(s) using techniques known in the art.
  • Vector components generally include, but are not limited to, one or more of a signal sequence, an origin of replication, one or more marker genes, and enhancer element, a promoter, and a transcription termination sequence. Construction of suitable vectors containing one or more of these components employs standard ligation techniques which are known to the skilled artisan.
  • Forms of the protein may be recovered from culture medium or from host cell lysates. If membrane-bound, it can be released from the membrane using a suitable detergent or through enzymatic cleavage. Cells employed for expression can also be disrupted by various physical or chemical means, such as freeze-thaw cycling, sonication, mechanical disruption or cell lysing agents.
  • Purification of the protein may be effected by any suitable technique known in the art, such as for example, fractionation on an ion-exchange column, ethanol precipitation, reverse phase HPLC, chromatography on silica or cation-exchange resin (e.g ., DEAE), chromatofocusing, SDS-PAGE, ammonium sulfate precipitation, gel filtration using protein A Sepharose columns (e.g ., Sephadex® G-75) to remove contaminants such as IgG, and metal chelating columns to bind epitope-tagged forms.
  • any suitable technique known in the art such as for example, fractionation on an ion-exchange column, ethanol precipitation, reverse phase HPLC, chromatography on silica or cation-exchange resin (e.g ., DEAE), chromatofocusing, SDS-PAGE, ammonium sulfate precipitation, gel filtration using protein A Sepharose columns (e.g ., Sephadex® G-75) to remove contaminants
  • the protein of choice is an antibody.
  • Techniques for the production of antibodies including polyclonal, monoclonal, humanized, bispecific and heteroconjugate antibodies follow.
  • Polyclonal antibodies are generally raised in animals by multiple subcutaneous (sc) or intraperitoneal (ip) injections of the relevant antigen and an adjuvant. It may be useful to conjugate the relevant antigen to a protein that is immunogenic in the species to be immunized, e.g., keyhole limpet hemocyanin, serum albumin, bovine thyroglobulin, or soybean trypsin inhibitor.
  • a protein that is immunogenic in the species to be immunized e.g., keyhole limpet hemocyanin, serum albumin, bovine thyroglobulin, or soybean trypsin inhibitor.
  • adjuvants which may be employed include Freund's complete adjuvant and MPL-TDM adjuvant (monophosphoryl Lipid A, synthetic trehalose dicorynomycolate).
  • the immunization protocol may be selected by one skilled in the art without undue experimentation.
  • the animals are boosted with 1/5 to 1/10 the original amount of peptide or conjugate in Freund's complete adjuvant by subcutaneous injection at multiple sites. Seven to 14 days later the animals are bled and the serum is assayed for antibody titer. Animals are boosted until the titer plateaus. Preferably, the animal is boosted with the conjugate of the same antigen, but conjugated to a different protein and/or through a different cross-linking reagent. Conjugates also can be made in recombinant cell culture as protein fusions. Also, aggregating agents such as alum are suitably used to enhance the immune response.
  • Monoclonal antibodies are obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts.
  • the modifier "monoclonal" indicates the character of the antibody as not being a mixture of discrete antibodies.
  • the monoclonal antibodies may be made using the hybridoma method first described by Kohler et al., Nature, 256:495 (1975 ), or may be made by recombinant DNA methods ( U.S. Patent No. 4,816,567 ).
  • a mouse or other appropriate host animal such as a hamster
  • lymphocytes that produce or are capable of producing antibodies that will specifically bind to the protein used for immunization.
  • lymphocytes may be immunized in vitro. Lymphocytes then are fused with myeloma cells using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell ( Goding, Monoclonal Antibodies: Principles and Practice, pp.59-103 (Academic Press, 1986 ).
  • the immunizing agent will typically include the protein to be formulated. Generally either peripheral blood lymphocytes ("PBLs") are used if cells of human origin are desired, or spleen cells or lymph node cells are used if non-human mammalian sources are desired.
  • PBLs peripheral blood lymphocytes
  • the lymphoctyes are then fused with an immortalized cell line using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell.
  • a suitable fusing agent such as polyethylene glycol
  • the hybridoma cells thus prepared are seeded and grown in a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells.
  • a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells.
  • the culture medium for the hybridomas typically will include hypoxanthine, aminopterin, and thymidine (HAT medium), which substances prevent the growth of HGPRT-deficient cells.
  • Preferred myeloma cells are those that fuse efficiently, support stable high-level production of antibody by the selected antibody-producing cells, and are sensitive to a medium such as HAT medium.
  • preferred myeloma cell lines are murine myeloma lines, such as those derived from MOPC-21 and MPC-11 mouse tumors available from the Salk Institute Cell Distribution Center, San Diego, California USA, and SP-2 cells available from the American Type Culture Collection, Rockville, Maryland USA.
  • Human myeloma and mouse-human heteromyeloma cell lines also have been described for the production of human monoclonal antibodies ( Kozbor, J. Immunol., 133:3001 (1984 ); Brodeur et al., Monoclonal Antibody Production Techniques and Applications, pp. 51-63 (Marcel Dekker, Inc., New York, 1987 )).
  • Culture medium in which hybridoma cells are growing is assayed for production of monoclonal antibodies directed against the antigen.
  • the binding specificity of monoclonal antibodies produced by hybridoma cells is determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunoabsorbent assay (ELISA).
  • RIA radioimmunoassay
  • ELISA enzyme-linked immunoabsorbent assay
  • the binding affinity of the monoclonal antibody can, for example, be determined by the Scatchard analysis of Munson et al., Anal. Biochem., 107:220 (1980 ).
  • the clones may be subcloned by limiting dilution procedures and grown by standard methods (Goding, supra ) .
  • Suitable culture media for this purpose include, for example, D-MEM or RPMI-1640 medium.
  • the hybridoma cells may be grown in vivo as ascites tumors in an animal.
  • the immunizing agent will typically include the epitope protein to which the antibody binds.
  • PBLs peripheral blood lymphocytes
  • spleen cells or lymph node cells are used if non-human mammalian sources are desired.
  • the lymphocytes are then fused with an immortalized cell line using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell.
  • a suitable fusing agent such as polyethylene glycol
  • Immortalized cell lines are usually transformed mammalian cells, particularly myeloma cells of rodent, bovine and human origin. Usually, rat or mouse myelome cell lines are employed.
  • the hybridoma cells may be cultured in a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, immortalized cells.
  • a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, immortalized cells.
  • the parental cells lack the enzyme hypoxanthine guanine phosphoribosyl transferase (HGPRT or HPRT)
  • HGPRT or HPRT hypoxanthine guanine phosphoribosyl transferase
  • the culture medium for the hybridomas typically will include hypoxanthine, aminopterin and thymidine (“HAT medium”), which substances prevent the growth of HGPRT-deficient cells.
  • Preferred immortalized cell lines are those that fuse efficiently, support stable high level expression of antibody by the selected antibody-producing cells, and are sensitive to a medium such as HAT medium. More preferred immortalized cell lines are murine myeloma lines, which can be obtained, for instance, from the Salk Institute Cell Distribution Center, San Diego, California and the American Type Culture Collection, Rockville, Maryland. Human myeloma and mouse-human heteromyeloma cell lines also have been described for the production of human monoclonal antibodies. Kozbor, J. Immunol., 133:3001 (1984 ); Brodeur et al., Monoclonal Antibody Production Techniques and Applications, Marcel Dekker, Inc., New York, (1987) pp. 51-63 .
  • the culture medium in which the hybridoma cells are cultured can then be assayed for the presence of monoclonal antibodies directed against the protein to be formulated.
  • the binding specificity of monoclonal antibodies produced by the hybridoma cells is determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunoabsorbent assay (ELISA).
  • RIA radioimmunoassay
  • ELISA enzyme-linked immunoabsorbent assay
  • the binding affinity of the monoclonal antibody can, for example, be determined by the Scatchard analysis of Munson and Pollard, Anal. Biochem., 107:220 (1980 ).
  • the clones may be subcloned by limiting dilution procedures and grown by standard methods. Goding, supra. Suitable culture media for this purpose include, for example, Dulbecco's Modified Eagle's Medium and RPMI-1640 medium. Alternatively, the hybridoma cells may be grown in vivo as ascites in a mammal.
  • the monoclonal antibodies secreted by the subclones are suitably separated from the culture medium, ascites fluid, or serum by conventional immunoglobulin purification procedures such as, for example, protein A-Sepharose, hydroxylapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography.
  • DNA encoding the monoclonal antibodies is readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of murine antibodies).
  • the hybridoma cells serve as a preferred source of such DNA.
  • the DNA may be placed into expression vectors, which are then transfected into host cells such as E. coli cells, simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells.
  • antibodies can be isolated from antibody phage libraries generated using the techniques described in McCafferty et al., Nature, 348:552-554 (1990 ). Clackson et al., Nature, 352:624-628 (1991 ) and Marks et al., J. Mol. Biol., 222:581-597 (1991 ) describe the isolation of murine and human antibodies, respectively, using phage libraries.
  • the DNA also may be modified, for example, by substituting the coding sequence for human heavy- and light-chain constant domains in place of the homologous murine sequences ( U.S. Patent No. 4,816,567 ; Morrison, et al., Proc. Natl Acad. Sci. USA, 81:6851 (1984 )), or by covalently joining to the immunoglobulin coding sequence all or part of the coding sequence for a non-immunoglobulin polypeptide.
  • non-immunoglobulin polypeptides are substituted for the constant domains of an antibody, or they are substituted for the variable domains of one antigen-combining site of an antibody to create a chimeric bivalent antibody comprising one antigen-combining site having specificity for an antigen and another antigen-combining site having specificity for a different antigen.
  • Chimeric or hybrid antibodies also may be prepared in vitro using known methods in synthetic protein chemistry, including those involving crosslinking agents.
  • immunotoxins may be constructed using a disulfide-exchange reaction or by forming a thioether bond.
  • suitable reagents for this purpose include iminothiolate and methyl-4-mercaptobutyrimidate.
  • the antibodies subject to the formulation method may further comprise humanized or human antibodies.
  • Humanized forms of non-human (e.g ., murine) antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab', F(ab') 2 or other antigen-binding subsequences of antibodies) which contain minimal sequence derived from non-human immunoglobulin.
  • Humanized antibodies include human immunoglobulins (recipient antibody) in which residues from a complementarity determining region (CDR) of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat or rabbit having the desired specificity, affinity and capacity.
  • CDR complementarity determining region
  • Fv framework residues of the human immunoglobulin are replaced by corresponding non-human residues.
  • Humanized antibodies may also comprise residues which are found neither in the recipient antibody nor in the imported CDR or framework sequences.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domain, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence.
  • the humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • Fc immunoglobulin constant region
  • a humanized antibody has one or more amino acid residues introduced into it from a source which is non-human. These non-human amino acid residues are often referred to as "import" residues, which are typically taken from an "import” variable domain. Humanization can be essentially performed following the method of Winter and co-workers, Jones et al., Nature 321:522-525 (1986 ); Riechmann et al., Nature 332:323-327 (1988 ); Verhoeyen et al., Science 239:1534-1536 (1988 ), or through substituting rodent CDRs or CDR sequences for the corresponding sequences of a human antibody.
  • humanized antibodies are chimeric antibodies ( U.S. Patent No. 4,816,567 ), wherein substantially less than an intact human variable domain has been substituted by the corresponding sequence from a non-human species.
  • humanized antibodies are typically human antibodies in which some CDR residues and possibly some FR residues are substituted by residues from analogous sites in rodent antibodies.
  • variable domains both light and heavy
  • sequence of the variable domain of a rodent antibody is screened against the entire library of known human variable-domain sequences.
  • the human sequence which is closest to that of the rodent is then accepted as the human framework (FR) for the humanized antibody.
  • FR human framework
  • Another method uses a particular framework derived from the consensus sequence of all human antibodies of a particular subgroup of light or heavy chains. The same framework may be used for several different humanized antibodies. Carter et al., Proc. Natl. Acad. Sci. USA, 89:4285 (1992 ); Presta et al., J. Immnol., 151:2623 (1993 ).
  • humanized antibodies are prepared by a process of analysis of the parental sequences and various conceptual humanized products using three-dimensional models of the parental and humanized sequences.
  • Three-dimensional immunoglobulin models are commonly available and are familiar to those skilled in the art.
  • Computer programs are available which illustrate and display probable three-dimensional conformational structures of selected candidate immunoglobulin sequences. Inspection of these displays permits analysis of the likely role of the residues in the functioning of the candidate immunoglobulin sequence, i.e., the analysis of residues that influence the ability of the candidate immunoglobulin to bind its antigen.
  • FR residues can be selected and combined from the recipient and import sequences so that the desired antibody characteristic, such as increased affinity for the target antigen(s), is achieved.
  • the CDR residues are directly and most substantially involved in influencing antigen binding.
  • transgenic animals e.g., mice
  • transgenic animals e.g., mice
  • J H antibody heavy-chain joining region
  • Human antibodies can also be derived from phage-display libraries ( Hoogenboom et al., J. Mol. Biol., 227:381 (1991 ); Marks et al., J. Mol. Biol., 222:581-597 (1991 )).
  • Human antibodies can also be produced using various techniques known in the art, including phage display libraries. Hoogenboom and Winter, J. Mol. Biol. 227: 381 (1991 ); Marks et al., J. Mol. Biol. 222: 581 (1991 ). The techniques of Cole et al., and Boerner et al., are also available for the preparation of human monoclonal antibodies ( Cole et al., Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, p. 77 (1985 ) and Boerner et al., J. Immunol. 147(1): 86-95 (1991 ).
  • human antibodies can be made by introducing human immunoglobulin loci into transgenic animals, e.g ., mice in which the endogenous immunoglobulin genes have been partially or completely inactivated. Upon challenge, human antibody production is observed, which closely resemble that seen in human in all respects, including gene rearrangement, assembly and antibody repertoire. This approach is described, for example, in U.S. Patent Nos.
  • ADPT Antibody Dependent Enzyme-Mediated Prodrug Therapy
  • the antibodies of the present invention may also be used in ADEPT by conjugating the antibody to a prodrug-activating enzyme which converts a prodrug (e.g . a peptidyl chemotherapeutic agent, see WO 81/01145 ) to an active anti-cancer drug.
  • a prodrug e.g . a peptidyl chemotherapeutic agent, see WO 81/01145
  • an active anti-cancer drug See, for example, WO 88/07378 and U. S. Patent No. 4,975,278 .
  • the enzyme component of the immunoconjugate useful for ADEPT includes any enzyme capable of acting on a prodrug in such as way so as to convert it into its more active, cytotoxic form.
  • Enzymes that are useful in the method of this invention include, but are not limited to, glycosidase, glucose oxidase, human lysozyme, human glucuronidase, alkaline phosphatase useful for converting phosphate-containing prodrugs into free drugs; arylsulfatase useful for converting sulfate-containing prodrugs into free drugs; cytosine deaminase useful for converting non-toxic 5-fluorocytosine into the anti-cancer drug 5-fluorouracil; proteases, such as serratia protease, thermolysin, subtilisin, carboxypeptidases ( e.g ., carboxypeptidase G2 and carboxypeptidase A) and cathepsins (such as cathepsins B and L), that are useful for converting peptide-containing prodrugs into free drugs; D-alanylcarboxypeptidases, useful for converting prodrugs that contain D-amin
  • antibodies with enzymatic activity can be used to convert the prodrugs of the invention into free active drugs (see, e.g., Massey, Nature 328: 457-458 (1987 )).
  • Antibody-abzyme conjugates can be prepared as described herein for delivery of the abzyme to a tumor cell population.
  • the enzymes of this invention can be covalently bound to the anti-IL-17 or anti-LIF antibodies by techniques well known in the art such as the use of the heterobifunctional cross-linking agents discussed above.
  • fusion proteins comprising at least the antigen binding region of the antibody of the invention linked to at least a functionally active portion of an enzyme of the invention can be constructed using recombinant DNA techniques well known in the art (see, e.g. Neuberger et al., Nature 312: 604-608 (1984 )).
  • Bispecific antibodies are antibodies that have binding specificities for at least two different epitopes. Such antibodies can be derived from full length antibodies or antibody fragments ( e.g. F(ab') 2 bispecific antibodies).
  • bispecific antibodies are known in the art. Traditional production of full length bispecific antibodies is based on the coexpression of two immunoglobulin heavy chain-light chain pairs, where the two chains have different specificities. Millstein et al., Nature, 305:537-539 (1983 ). Because of the random assortment of immunoglobulin heavy and light chains, these hybridomas (quadromas) produce a potential mixture of 10 different antibody molecules, of which only one has the correct bispecific structure. Purification of the correct molecule, which is usually done by affinity chromatography steps, is rather cumbersome, and the product yields are low. Similar procedures are disclosed in WO 93/08829 and in Traunecker et al., EMBO J., 10:3655-3659 (1991 ).
  • Antibody variable domains with the desired binding specificities can be fused to immunoglobulin constant domain sequences.
  • the fusion preferably is with an immunoglobulin heavy-chain constant domain, comprising at least part of the hinge, CH2, and CH3 regions. It is preferred to have the first heavy-chain constant region (CH1) containing the site necessary for light-chain binding present in at least one of the fusions.
  • DNAs encoding the immunoglobulin heavy-chain fusions, and, if desired, the immunoglobulin light chain are inserted into separate expression vectors, and are co-transfected into a suitable host organism.
  • antibody variable domains with the desired binding specificities are fused to immunoglobulin constant domain sequences.
  • the fusion preferably is with an immunoglobulin heavy chain constant domain, comprising at least part of the hinge, CH2, and CH3 regions. It is preferred to have the first heavy-chain constant region (CH1) containing the site necessary for light chain binding, present in at least one of the fusions.
  • DNAs encoding the immunoglobulin heavy chain fusions and, if desired, the immunoglobulin light chain are inserted into separate expression vectors, and are co-transfected into a suitable host organism.
  • the interface between a pair of antibody molecules can be engineered to maximize the percentage of heterodimers which are recovered from recombinant cell culture.
  • the preferred interface comprises at least a part of the CH3 region of an antibody constant domain.
  • one or more small amino acid side chains from the interface of the first antibody molecule are replaced with larger side chains (e.g., tyrosine or tryptophan).
  • Compensatory "cavities" of identical or similar size to the large side chains(s) are created on the interface of the second antibody molecule by replacing large amino acid side chains with smaller ones (e.g ., alanine or threonine). This provides a mechanism for increasing the yield of the heterodimer over other unwanted end-products such as homodimers.
  • the bispecific antibodies are composed of a hybrid immunoglobulin heavy chain with a first binding specificity in one arm, and a hybrid immunoglobulin heavy chain-light chain pair (providing a second binding specificity) in the other arm. It was found that this asymmetric structure facilitates the separation of the desired bispecific compound from unwanted immunoglobulin chain combinations, as the presence of an immunoglobulin light chain in only one half of the bispecific molecule provides for a facile way of separation. This approach is disclosed in WO 94/04690, published March 3, 1994 . For further details of generating bispecific antibodies see, for example, Suresh et al., Methods in Enzymology, 121:210 (1986 ).
  • Bispecific antibodies include cross-linked or "heteroconjugate" antibodies.
  • one of the antibodies in the heteroconjugate can be coupled to avidin, the other to biotin.
  • Such antibodies have, for example, been proposed to target immune system cells to unwanted cells ( US Patent No. 4,676,980 ), and for treatment of HIV infection ( WO 91/00360 , WO 92/200373 ).
  • Heteroconjugate antibodies may be made using any convenient cross-linking methods. Suitable cross-linking agents are well known in the art, and are disclosed in US Patent No. 4,676,980 , along with a number of cross-linking techniques.
  • bispecific antibodies from antibody fragments
  • the following techniques can also be used for the production of bivalent antibody fragments which are not necessarily bispecific.
  • Fab' fragments recovered from E. coli can be chemically coupled in vitro to form bivalent antibodies. See, Shalaby et al., J. Exp. Med., 175:217-225 (1992 ).
  • Bispecific antibodies can be prepared as full length antibodies or antibody fragments (e.g . F(ab') 2 bispecific antibodies). Techniques for generating bispecific antibodies from antibody fragments have been described in the literature. For example, bispecific antibodies can be prepared using chemical linkage. Brennan et al., Science 229: 81 (1985 ) describe a procedure wherein intact antibodies are proteolytically cleaved to generate F(ab') 2 fragments. These fragments are reduced in the presence of the dithiol complexing agent sodium arsenite to stabilize vicinal dithiols and prevent intermolecular disulfide formation. The Fab' fragments generated are then converted to thionitrobenzoate (TNB) derivatives. One of the Fab'-TNB derivatives is then reconverted to the Fab'-TNB derivative to form the bispecific antibody.
  • the bispecific antibodies produced can be used as agents for the selective immobilization of enzymes.
  • Fab' fragments may be directly recovered from E. coli and chemically coupled to form bispecific antibodies.
  • Shalaby et al., J. Exp. Med. 175: 217-225 (1992 ) describes the production of fully humanized bispecific antibody F(ab') 2 molecules.
  • Each Fab' fragment was separately secreted from E. coli and subjected to directed chemical coupling in vitro to form the bispecific antibody.
  • the bispecific antibody thus formed was able to bind to cells overexpressing the ErbB2 receptor and normal human T cells, as well as trigger the lytic activity of human cytotoxic lymphocytes against human breast tumor targets.
  • bivalent heterodimers have been produced using leucine zippers.
  • the leucine zipper peptides from the Fos and Jun proteins were linked to the Fab' portions of two different antibodies by gene fusion.
  • the antibody homodimers were reduced at the hinge region to form monomers and then re-oxidized to form the antibody heterodimers.
  • the "diabody" technology described by Hollinger et al., Proc. Natl. Acad. Sci.
  • the fragments comprise a heavy-chain variable domain (V H ) connected to a light-chain variable domain (V L ) by a linker which is too short to allow pairing between the two domains on the same chain. Accordingly, the V H and V L domains of one fragment are forced to pair with the complementary V L and V H domains of another fragment, thereby forming two antigen-binding sites.
  • V H and V L domains of one fragment are forced to pair with the complementary V L and V H domains of another fragment, thereby forming two antigen-binding sites.
  • sFv single-chain Fv
  • Antibodies with more than two valencies are contemplated.
  • trispecific antibodies can be prepared. Tutt et al., J. Immunol. 147: 60 (1991 ).
  • bispecific antibodies may bind to two different epitopes on a given molecule.
  • an anti-protein arm may be combined with an arm which binds to a triggering molecule on a leukocyte such as a T-cell receptor molecule (e.g ., CD2, CD3, CD28 or B7), or Fc receptors for IgG (Fc ⁇ R), such as Fc ⁇ RI (CD64), Fc ⁇ RII (CD32) and Fc ⁇ RIII (CD16) so as to focus cellular defense mechanisms to the cell expressing the particular protein.
  • Bispecific antibocis may also be used to localize cytotoxic agents to cells which express a particular protein.
  • Such antibodies possess a protein-binding arm and an arm which binds a cytotoxic agent or a radionuclide chelator, such as EOTUBE, DPTA, DOTA or TETA.
  • a cytotoxic agent or a radionuclide chelator such as EOTUBE, DPTA, DOTA or TETA.
  • Another bispecific antibody of interest binds the protein of interest and further binds tissue factor (TF).
  • Heteroconjugate antibodies are also within the scope of the present invention.
  • Heteroconjugate antibodies are composed of two covalently joined antibodies. Such antibodies have, for example, been proposed to tatget immune system cells to unwanted cells, U.S.P. 4,676,980 , and for treatment of HIV infection. WO 91/00360 , WO 92/200373 and EP 03089 .
  • the antibodies may be prepared in vitro using known methods in synthetic protein chemistry, including those involving crosslinking agents.
  • immunotoxins may be constructed using a disulfide exchange reaction or by forming a thioether bond. Examples of suitable reagents for this purpose include iminothiolate and methyl-4-mercaptobutyrimidate and those disclosed, for example, in U.S. Patent No. 4,676,980 .
  • the target polypeptide When the target polypeptide is expressed in a recombinant cell other than one of human origin, the target polypeptide is completely free of proteins or polypeptides of human origin. However, it is necessary to purify the target polypeptide from recombinant cell proteins or polypeptides to obtain preparations that are substantially homogeneous as to the target polypeptide.
  • the culture medium or lysate is typically centrifuged to remove particulate cell debris. The membrane and soluble protein fractions are then separated. The target polypeptide may then be purified from the soluble protein fraction and from the membrane fraction of the culture lysate, depending on whether the target polypeptide is membrane bound.
  • the following procedures are exemplary of suitable purification procedures: fractionation on immunoaffinity or ion-exchange columns; ethanol precipitation; reverse phase HPLC; chromatography on silica or on a cation exchange resin such as DEAE; chromatofocusing; SDS-PAGE; ammonium sulfate precipitation; gel filtration using, for example, Sephadex G-75; and protein A Sepharose columns to remove contaminants such as IgG.
  • MAbs monoclonal antibodies
  • HCP host cell protein
  • HIC hydrophobic interaction chromatography
  • viruses that may be present in protein solutions are larger than the proteins themselves. It is thus presumed that viruses can be removed from proteins in accordance with size, by filtration.
  • Virus filtration can remove larger, e.g., retroviruses (80 - 100 nm diameter), typically using high throughput membranes with nominal pore size of about 60 nm. Since high throughput membranes with nominal pore size of 20 nm are also commercially available, it is possible to remove smaller viruses by filtration, such as, for example, parvoviruses (18 - 26 nm diameter), while allowing passage of proteins that are as large as 160 kD ( ⁇ 8 nm), e.g., monoclonal antibodies.
  • the present invention is primarily intended for resolving issues typically associated with the filtration of such smaller viruses, using viral removal filters of smaller pore size.
  • virus filtration step can be implemented at any one of several points in a given downstream process.
  • virus filtration may take place following a low pH viral inactivation step, or following an intermediate column chromatography step, or after a final column chromatography step.
  • virus filtration unit operation could be carried out at any stage in the downstream process.
  • Virus filtration during downstream processing of monoclonal antibody is typically performed after an affinity chromatography step (capture step) and an ion-exchange purification step (polishing step).
  • the protein solution In tangential flow virus filtration, the protein solution is usually pumped around at a constant rate of flow on the retention side.
  • the differential pressure generated across the virus removal filter allows protein solution to permeate through the filter while the viruses are retained on the retentate side.
  • normal-flow or "dead-end” virus filtration
  • the same virus filter as that used in tangential virus filtration can be used, although the peripheral equipment and operating procedures are much simpler and less expensive than in the case of tangential flow virus filtration.
  • "normal-flow” filtration involves placing the macromolecule-containing solution in a pressure vessel prior to filtration and pressing the solution through the virus removal filter with the aid of a pressure source, suitably nitrogen (gas) or air.
  • a pressure source suitably nitrogen (gas) or air.
  • a pump could be used on the retentate side to filter the liquid through the virus removal filter at a pre-determined flow rate.
  • the degree of fineness of filters generally, is normally expressed as pore size or the approximate molecular weight (relative molecular mass) at which the molecules are stopped by the filter, the so called cut-off.
  • Virus filters are known in the art and are supplied by Millipore from Massachusetts, USA and Asahi Chemical Industry Co., Ltd. from Japan, among others. Suitable parvovirus retentive filters include Viresolve® Pro (Millipore Corp., Billerica, MA) Viresolve® Pro membrane has an asymmetric dual layer structure and is made from polyethersulfone (PES). The membrane structure is designed to retain viruses greater than 20 nm in size while allowing proteins of molecular weight less than 180 kDa to permeate through the membrane.
  • Viresolve® Pro Millipore Corp., Billerica, MA
  • PES polyethersulfone
  • the Novasip DV50 grade capsule filter incorporates an Ultipor VF DV50 grade Ultipleat® membrane cartridge for removal of viruses 40 - 50 nm and larger.
  • Novasip Ultipor VF capsule filters are supplied non-sterile and can also be Gamma-irradiated.
  • Virosart® CPV utilizes double - layer polyethersulfone asymmetric membrane and retains more than 4 log of parvoviruses and 6 log of retroviruses.
  • Prefiltration of the feed solution can have a dramatic impact on filter performance.
  • Prefiltration typically is targeted to remove impurities and contaminants that might lead to fouling of virus filters, such as protein aggregates, DNA and other trace materials.
  • a striking enhancement of the efficacy of virus filters can be achieved by a prefiltration step including the use of both cation exchange and endotoxin removal media.
  • the term “medium” or “media” is used to cover any means for performing the cation exchange and endotoxin removal steps, respectively.
  • the term “cation exchange medium” specifically includes, without limitation, cation exchange resins, matrices, absorbers, and the like.
  • endotoxin removal medium includes, without limitation, any positively charged membrane surface, including, for example, chromatographic endotoxin removal media, endotoxin affinity removal media, and the like.
  • Cation exchange media suitable for use in the prefiltration step of the present invention include, without limitation, Mustang® S, Sartobind® S, Viresolve® Shield, SPFF, SPXL, Capto® S, Poros® 50 HS, Fractogel® S, Hypercel® D etc., which are commercially available.
  • Endotoxin removal media suitable for use in the prefiltration step of the present invention include, without limitation, Mustang® E, Mustang® Q, Sartobind® Q, Chromasorb®, Possidyne®, Capto® Q, QSFF, Poros® Q, Fractogel® Q etc., which are commercially available.
  • the pre-filtration step can be performed, for example, by taking the in process chromatography pool and processing the pool over a filtration train that comprises the endotoxin removal and cation exchange media and parvovirus filter.
  • the endotoxin removal and cation exchange media act as pre-filtration steps and the capacity of parvovirus filter is independent of the sequence of two steps in the filtration train.
  • the filtration train can work continuously as a single step or it can be operated as different unit operations. For example, in one embodiment, the chromatography pool is first processed over endotoxin removal media, the collected pool is then processed over cation exchange media and the subsequent pool is filtered with parvovirus filter.
  • the order of applying the cation exchange media and endotoxin removal media in the process sequence does not impact parvovirus filtration capacity.
  • the process can be operated over a wide pH range, such as, for example, in the pH range of 4 - 10, with optimal filter capacity being dependent on the target impurity profile and product attributes.
  • protein concentrations can vary over a wide range, such as, for example, 1 - 40 g/L, and does not limit the mass throughput of parvovirus filters.
  • Protein concentration was determined using a UV-vis spectrophotometer (NanoDrop ND - 1000, NanoDrop Technologies, Wilmington, DE) with absorbance measured at 280 nm.
  • Viresolve® Pro (Millipore Corp., Billerica, MA) parvovirus retentive filter.
  • Viresolve® Pro membrane has an asymmetric dual layer structure and is made from polyethersulfone (PES). The membrane structure is designed to retain viruses greater than 20 nm in size while allowing proteins of molecular weight less than 180 kDa to permeate through the membrane.
  • PES polyethersulfone
  • Viresolve® Pro evaluated in this study included Viresolve® Optiscale 40 depth filter (Millipore Corp., Billerica, MA), Fluorodyne EX Mini 0.2 ⁇ m sterile filter (Pall Corp., East Hills, NY) and the membrane adsorbers from Mustang® family (Pall Corp., East Hills, NY).
  • the membrane adsorbers were procured from the vendor in fully encapsulated Acrodisc® units. Table 1 summarizes the key properties (functional group, bed volume, pore size etc.) of all the pre-filters used in this study.
  • Downstream purification of mAbs expressed in mammalian cell cultures typically utilize centrifugation and depth filtration as a first step to remove cells and cell debris, followed by affinity chromatography for mAb capture and removal of host cell proteins (HCP), followed by cation exchange chromatography, virus filtration, and anion exchange chromatography for further removal of impurities such as aggregates, viruses, leached protein A and HCP's.
  • HCP host cell proteins
  • Figure 2 shows the experimental data for differential pressure across Viresolve Pro at a constant flux of 200 L/m 2 /hr with a therapeutic mAb feed stream with different prefilters.
  • X-axis represents the mass of mAb loaded per square meter of virus filter.
  • Y-axis represents the differential pressure across the virus filter as a function of mass throughput. The data clearly indicates that the depth filter provides several orders of magnitude increase in virus filtration capacity compared to sterile filter. Similar observations were made by Bolton et al. ( Bolton et al. Appl. Biochem.
  • depth filters have traditionally been used successfully for clarification of cell culture fluid, there are quite a few limitations that deserve extra consideration when used downstream of capture steps, e.g., as a prefilter to parvovirus retentive filter.
  • Endotoxin removal media by itself can effectively increase the capacity of parvovirus filters when used for prefiltration and (2) coupling of endotoxin removal and cation exchange media in the prefiltration train can provide several-fold increase in parvovirus filtration capacity, lowering raw material costs and facilitating successful operation of virus filtration at manufacturing scale.
EP17195123.9A 2009-08-06 2010-08-06 Procédé pour améliorer l'élimination de virus dans la purification de protéines Pending EP3309168A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US23181109P 2009-08-06 2009-08-06
EP10740823.9A EP2462158B1 (fr) 2009-08-06 2010-08-06 Methode pour ameliorer l'elimination de virus pendant la purification de proteines

Related Parent Applications (3)

Application Number Title Priority Date Filing Date
EP10740823.9A Division EP2462158B1 (fr) 2009-08-06 2010-08-06 Methode pour ameliorer l'elimination de virus pendant la purification de proteines
EP10740823.9A Division-Into EP2462158B1 (fr) 2009-08-06 2010-08-06 Methode pour ameliorer l'elimination de virus pendant la purification de proteines
PCT/US2010/044760 Previously-Filed-Application WO2011031397A1 (fr) 2009-08-06 2010-08-06 Procédé permettant d'améliorer l'élimination d'un virus lors de la purification des protéines

Publications (1)

Publication Number Publication Date
EP3309168A1 true EP3309168A1 (fr) 2018-04-18

Family

ID=42985499

Family Applications (2)

Application Number Title Priority Date Filing Date
EP10740823.9A Active EP2462158B1 (fr) 2009-08-06 2010-08-06 Methode pour ameliorer l'elimination de virus pendant la purification de proteines
EP17195123.9A Pending EP3309168A1 (fr) 2009-08-06 2010-08-06 Procédé pour améliorer l'élimination de virus dans la purification de protéines

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP10740823.9A Active EP2462158B1 (fr) 2009-08-06 2010-08-06 Methode pour ameliorer l'elimination de virus pendant la purification de proteines

Country Status (25)

Country Link
US (4) US10662237B2 (fr)
EP (2) EP2462158B1 (fr)
JP (5) JP2013501075A (fr)
KR (3) KR20190018041A (fr)
CN (2) CN102712691A (fr)
AU (1) AU2010292897B2 (fr)
BR (1) BR112012002574A2 (fr)
CA (1) CA2770235C (fr)
DK (1) DK2462158T3 (fr)
ES (1) ES2662529T3 (fr)
HR (1) HRP20180309T1 (fr)
HU (1) HUE038451T2 (fr)
IL (3) IL217740B (fr)
LT (1) LT2462158T (fr)
MX (1) MX346115B (fr)
MY (1) MY188566A (fr)
NO (1) NO2462158T3 (fr)
NZ (1) NZ597809A (fr)
PL (1) PL2462158T3 (fr)
PT (1) PT2462158T (fr)
RU (1) RU2573894C2 (fr)
SG (1) SG178276A1 (fr)
SI (1) SI2462158T1 (fr)
WO (1) WO2011031397A1 (fr)
ZA (1) ZA201200626B (fr)

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8911964B2 (en) 2006-09-13 2014-12-16 Abbvie Inc. Fed-batch method of making human anti-TNF-alpha antibody
CN103555652A (zh) 2006-09-13 2014-02-05 Abbvie公司 细胞培养改良
TWI610936B (zh) 2008-10-20 2018-01-11 艾伯維有限公司 使用蛋白質a親和性層析進行抗體之分離及純化
CA2738499A1 (fr) 2008-10-20 2010-04-29 Abbott Laboratories Inactivation virale pendant la purification d'anticorps
PL2462158T3 (pl) 2009-08-06 2018-05-30 F. Hoffmann-La Roche Ag Sposób poprawy usuwania wirusów przy oczyszczaniu białka
TW201302793A (zh) 2010-09-03 2013-01-16 Glaxo Group Ltd 新穎之抗原結合蛋白
BR112013024521A2 (pt) 2011-03-25 2019-09-24 Genentech Inc métodos de purificação de proteínas
DE102011105525B4 (de) * 2011-06-24 2015-03-26 Sartorius Stedim Biotech Gmbh Verfahren zur Abtrennung von Biopolymer-Aggregaten und Viren aus einem Fluid
WO2013028330A2 (fr) * 2011-08-19 2013-02-28 Emd Millipore Corporation Procédés pour réduire le niveau d'une ou de plusieurs impuretés dans un échantillon pendant un procédé de purification de protéines
SG11201403437TA (en) 2011-12-22 2014-07-30 Genentech Inc Ion exchange membrane chromatography
KR102571391B1 (ko) 2013-09-13 2023-08-29 제넨테크, 인크. 정제된 재조합 폴리펩티드를 포함하는 방법 및 조성물
PL3044323T3 (pl) 2013-09-13 2022-06-27 F.Hoffmann-La Roche Ag Sposoby wykrywania i ilościowego oznaczania białka komórki gospodarza w liniach komórkowych
LT3209767T (lt) * 2014-10-21 2020-11-25 Gennova Biopharmaceuticals Ltd. Naujas gryninimo būdas, skirtas rekombinantinės tnk-tpa (tenekteplazės) išskyrimui ir pramoninei gamybai
WO2016154290A1 (fr) 2015-03-23 2016-09-29 Alexion Pharmaceuticals, Inc. Filtration virale
CN110740802B (zh) * 2017-06-12 2022-04-15 旭化成医疗株式会社 含有蛋白质的溶液的过滤方法
SG11202005809TA (en) * 2017-12-21 2020-07-29 Genzyme Corp Methods for enhanced removal of impurities during protein a chromatography

Citations (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0003089A1 (fr) 1978-01-06 1979-07-25 Bernard David Séchoir pour feuilles imprimées par sérigraphie
WO1981001145A1 (fr) 1979-10-18 1981-04-30 Univ Illinois Medicaments "pro-drugs" pouvant etre actives par des enzymes hydrolytiques
US4399216A (en) 1980-02-25 1983-08-16 The Trustees Of Columbia University Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
US4676980A (en) 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
WO1988007378A1 (fr) 1987-03-09 1988-10-06 Cancer Research Campaign Technology Ltd. Amelioration apportees a des systemes d'administration de medicaments
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
WO1989005859A1 (fr) 1987-12-21 1989-06-29 The Upjohn Company Transformation par l'agrobacterium de graines de plantes de germination
US4975278A (en) 1988-02-26 1990-12-04 Bristol-Myers Company Antibody-enzyme conjugates in combination with prodrugs for the delivery of cytotoxic agents to tumor cells
EP0404097A2 (fr) 1989-06-22 1990-12-27 BEHRINGWERKE Aktiengesellschaft Récepteurs mono- et oligovalents, bispécifiques et oligospécifiques, ainsi que leur production et application
WO1991000360A1 (fr) 1989-06-29 1991-01-10 Medarex, Inc. Reactifs bispecifiques pour le traitement du sida
WO1992020373A1 (fr) 1991-05-14 1992-11-26 Repligen Corporation Anticorps d'heteroconjugues pour le traitement des infections a l'hiv
WO1993008829A1 (fr) 1991-11-04 1993-05-13 The Regents Of The University Of California Compositions induisant la destruction de cellules infectees par l'hiv
WO1993011161A1 (fr) 1991-11-25 1993-06-10 Enzon, Inc. Proteines multivalentes de fixation aux antigenes
WO1994004690A1 (fr) 1992-08-17 1994-03-03 Genentech, Inc. Immunoadhesines bispecifiques
US5500362A (en) 1987-01-08 1996-03-19 Xoma Corporation Chimeric antibody with specificity to human B cell surface antigen
US5545807A (en) 1988-10-12 1996-08-13 The Babraham Institute Production of antibodies from transgenic animals
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
WO1996027011A1 (fr) 1995-03-01 1996-09-06 Genentech, Inc. Procede d'obtention de polypeptides heteromultimeriques
US5569825A (en) 1990-08-29 1996-10-29 Genpharm International Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5641870A (en) 1995-04-20 1997-06-24 Genentech, Inc. Low pH hydrophobic interaction chromatography for antibody purification
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5821337A (en) 1991-06-14 1998-10-13 Genentech, Inc. Immunoglobulin variants
US20030146156A1 (en) * 2002-02-04 2003-08-07 Martin Siwak Process for removing protein aggregates and virus from a protein solution
WO2007108955A1 (fr) * 2006-03-20 2007-09-27 Medarex, Inc. Purification de protéines
WO2009017491A1 (fr) * 2006-06-14 2009-02-05 Smithkline Beecham Corporation Procédés de purification d'anticorps à l'aide de céramique d'hydroxyapatite

Family Cites Families (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2058199T3 (es) 1987-09-23 1994-11-01 Bristol Myers Squibb Co Heteroconjugados de anticuerpos para la eliminacion de celulas infectadas por el vih.
JPH02198687A (ja) 1989-01-26 1990-08-07 Asahi Chem Ind Co Ltd 純水の製造方法
JPH038759A (ja) 1989-03-16 1991-01-16 Sumitomo Metal Ind Ltd 半導体磁器物質
CA2166805A1 (fr) * 1993-07-09 1995-01-19 Thomas M. Smith Purification des proteines
ES2126775T3 (es) 1993-09-21 1999-04-01 Hemosol Inc Procedimiento cromatografico de desplazamiento y producto de hemoglobina purificada.
CA2106612C (fr) 1993-09-21 2001-02-06 Diana Pliura Processus de chromatographie a deplacement
JP3415222B2 (ja) 1993-10-20 2003-06-09 東洋濾紙株式会社 エンドトキシン吸着能を有するポリエーテルサルホン又はスルホン化ポリエーテルサルホン微孔膜
JP3453623B2 (ja) 1994-03-30 2003-10-06 サンスター株式会社 エンドトキシン吸着除去剤および除去方法
WO1996003202A1 (fr) 1994-07-28 1996-02-08 Millipore Corporation Membrane composite poreuse et son procede de fabrication
FR2737730B1 (fr) * 1995-08-10 1997-09-05 Pasteur Merieux Serums Vacc Procede de purification de virus par chromatographie
US6750333B1 (en) * 1996-02-06 2004-06-15 Roche Diagnostics Gmbh Process for preparing purified nucleic acid and the use thereof
TW505655B (en) 1997-10-14 2002-10-11 Tanox Inc Enhanced aggregate removal from bulk-biologicals using ion exchange chromatography
GB9907553D0 (en) 1999-04-01 1999-05-26 Cantab Pharma Res Purification of biological preparations
EP1200179B8 (fr) 1999-07-30 2007-02-21 Genentech, Inc. Membranes de filtration chargees et utilisations associees
ES2184594B1 (es) 2001-01-17 2004-01-01 Probitas Pharma Sa Procedimiento para la produccion de gammaglobulina g humana inactivada de virus.
US6806355B2 (en) 2001-08-14 2004-10-19 Statens Serum Institut Purification process for large scale production of Gc-globulin, the Gc-globulin produced hereby, a use of Gc.globulin and a Gc-globulin medicinal product
SI1501369T1 (sl) 2002-04-26 2015-10-30 Genentech, Inc. Neafinitetno čiščenje proteinov
CA2498725C (fr) 2002-09-13 2014-07-08 Biogen Idec Inc. Procede de purification de polypeptides par chromatographie sur lit mobile simule
EP1403274A1 (fr) 2002-09-30 2004-03-31 Meristem Therapeutics Procédé de purification de protéines recombinantes d'un milieu complexe et protéines purifiées par ce procédé
US20040116676A1 (en) 2002-09-30 2004-06-17 Hotta Joann Methods for removal of contaminants from blood product solutions
GB0304576D0 (en) 2003-02-28 2003-04-02 Lonza Biologics Plc Protein a chromatography
JPWO2004087761A1 (ja) 2003-03-31 2006-07-27 麒麟麦酒株式会社 ヒトモノクローナル抗体およびヒトポリクローナル抗体の精製
CA2521826C (fr) 2003-04-11 2013-08-06 Jennifer L. Reed Anticorps il-9 recombinants et leurs utilisations
LT2418220T (lt) * 2003-12-10 2017-10-10 E. R. Squibb & Sons, L.L.C. Interferono-alfa antikūnai ir jų panaudojimas
BRPI0507298A (pt) 2004-01-30 2007-07-03 Suomen Punainen Risti Veripalv processo para a produção de imunoglobulina segura a vìrus
EP1713904B1 (fr) 2004-01-30 2016-06-29 Shire Pharmaceuticals Ireland Limited Production et purification d'arylsulfatase a recombinante
US7390403B2 (en) 2004-03-19 2008-06-24 Millipore Corporation Prefilter system for biological systems
EP1888208B1 (fr) 2004-10-21 2012-11-14 Statens Serum Institut Procede incluant la nanofiltration permettant d'obtenir un produit mbl resistant aux agents infectieux
EP1858928A2 (fr) * 2005-03-08 2007-11-28 Pharmacia & Upjohn Company LLC Compositions d'anticorps anti-m-csf a niveaux d'endotoxine reduits
EP1854810A1 (fr) 2006-05-09 2007-11-14 PanGenetics B.V. Anticorps monoclonal anti CD40 humain déimmunisé et antagoniste dérivé de l'anticorps ch5D12
CA2661872A1 (fr) 2006-08-28 2008-03-06 Ares Trading S.A. Procede de purification de proteines fc-hybrides
WO2008036899A2 (fr) 2006-09-22 2008-03-27 Amgen Inc. Procédés d'élimination des contaminants viraux durant la purification des protéines
RU2460541C2 (ru) 2006-10-27 2012-09-10 Лпат, Инк. Композиции и способы связывания сфингозин-1-фосфата
WO2008073620A2 (fr) 2006-11-02 2008-06-19 Neose Technologies, Inc. Procédé de fabrication pour la production de polypeptides exprimés dans des lignées cellulaires d'insectes
CA2668771C (fr) 2006-11-08 2016-03-15 Wyeth Milieux concus rationnellement pour une culture cellulaire
ES2624185T3 (es) 2007-04-26 2017-07-13 Chugai Seiyaku Kabushiki Kaisha Método de cultivo celular que utiliza un medio enriquecido con aminoácidos
CA2708951A1 (fr) 2007-12-21 2009-07-09 Genetech, Inc. Cristallisation d'anticorps anti-cd20
US20110105725A1 (en) 2008-05-15 2011-05-05 Novo Nordisk A/S Antibody purification process
EP2412817B2 (fr) * 2009-03-27 2019-06-05 Asahi Kasei Medical Co., Ltd. Procédé pour éliminer des virus dans une solution d'anticorps monoclonal à haute concentration
AU2010253830B2 (en) 2009-05-27 2016-03-03 Takeda Pharmaceutical Company Limited A method to produce a highly concentrated immunoglobulin preparation for subcutaneous use
PL2462158T3 (pl) 2009-08-06 2018-05-30 F. Hoffmann-La Roche Ag Sposób poprawy usuwania wirusów przy oczyszczaniu białka
JP5787891B2 (ja) 2009-09-01 2015-09-30 ジェネンテック, インコーポレイテッド 改変されたプロテインa溶離による向上したタンパク質精製

Patent Citations (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0003089A1 (fr) 1978-01-06 1979-07-25 Bernard David Séchoir pour feuilles imprimées par sérigraphie
WO1981001145A1 (fr) 1979-10-18 1981-04-30 Univ Illinois Medicaments "pro-drugs" pouvant etre actives par des enzymes hydrolytiques
US4399216A (en) 1980-02-25 1983-08-16 The Trustees Of Columbia University Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4676980A (en) 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
US5500362A (en) 1987-01-08 1996-03-19 Xoma Corporation Chimeric antibody with specificity to human B cell surface antigen
WO1988007378A1 (fr) 1987-03-09 1988-10-06 Cancer Research Campaign Technology Ltd. Amelioration apportees a des systemes d'administration de medicaments
WO1989005859A1 (fr) 1987-12-21 1989-06-29 The Upjohn Company Transformation par l'agrobacterium de graines de plantes de germination
US4975278A (en) 1988-02-26 1990-12-04 Bristol-Myers Company Antibody-enzyme conjugates in combination with prodrugs for the delivery of cytotoxic agents to tumor cells
US5545807A (en) 1988-10-12 1996-08-13 The Babraham Institute Production of antibodies from transgenic animals
EP0404097A2 (fr) 1989-06-22 1990-12-27 BEHRINGWERKE Aktiengesellschaft Récepteurs mono- et oligovalents, bispécifiques et oligospécifiques, ainsi que leur production et application
WO1991000360A1 (fr) 1989-06-29 1991-01-10 Medarex, Inc. Reactifs bispecifiques pour le traitement du sida
US5569825A (en) 1990-08-29 1996-10-29 Genpharm International Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
WO1992020373A1 (fr) 1991-05-14 1992-11-26 Repligen Corporation Anticorps d'heteroconjugues pour le traitement des infections a l'hiv
US5821337A (en) 1991-06-14 1998-10-13 Genentech, Inc. Immunoglobulin variants
WO1993008829A1 (fr) 1991-11-04 1993-05-13 The Regents Of The University Of California Compositions induisant la destruction de cellules infectees par l'hiv
WO1993011161A1 (fr) 1991-11-25 1993-06-10 Enzon, Inc. Proteines multivalentes de fixation aux antigenes
WO1994004690A1 (fr) 1992-08-17 1994-03-03 Genentech, Inc. Immunoadhesines bispecifiques
WO1996027011A1 (fr) 1995-03-01 1996-09-06 Genentech, Inc. Procede d'obtention de polypeptides heteromultimeriques
US5641870A (en) 1995-04-20 1997-06-24 Genentech, Inc. Low pH hydrophobic interaction chromatography for antibody purification
US20030146156A1 (en) * 2002-02-04 2003-08-07 Martin Siwak Process for removing protein aggregates and virus from a protein solution
US7118675B2 (en) 2002-02-04 2006-10-10 Millipore Corporation Process for removing protein aggregates and virus from a protein solution
WO2007108955A1 (fr) * 2006-03-20 2007-09-27 Medarex, Inc. Purification de protéines
WO2009017491A1 (fr) * 2006-06-14 2009-02-05 Smithkline Beecham Corporation Procédés de purification d'anticorps à l'aide de céramique d'hydroxyapatite

Non-Patent Citations (87)

* Cited by examiner, † Cited by third party
Title
"Basic and Clinical Immunology", 1994, APPLETON & LANGE, pages: 71
"Mammalian Cell Biotechnology: A Practical Approach", 1991, IRL PRESS
"Pluckthun in The Pharmacology of Monoclonal Antibodies", vol. 113, 1994, SPRINGER-VERLAG, pages: 269 - 315
"Scatchard analysis of Munson and Pollard", ANAL. BIOCHEM., vol. 107, 1980, pages 220
BOERNER ET AL., J. IMMUNOL., vol. 147, no. 1, 1991, pages 86 - 95
BOHONAK; ZYDNEY, JOURNAL OF MEMBRANE SCIENCE, vol. 254, no. 1-2, 2005, pages 71 - 79
BOLTON ET AL., APPL. BIOCHEM., vol. 43, 2006, pages 55 - 63
BOLTON ET AL., BIOTECHNOL. APPL. BIOCHEM., vol. 43, 2006, pages 55 - 63
BOLTON ET AL., BIOTECHNOLOGY AND APPLIED BIOCHEMISTRY, vol. 42, 2005, pages 133 - 142
BRENNAN ET AL., SCIENCE, vol. 229, 1985, pages 81
BRODEUR ET AL.: "Monoclonal Antibody Production Techniques and Applications", 1987, MARCEL DEKKER, INC., pages: 51 - 63
BROWN ET AL., IBC'S 20TH ANTIBODY DEVELOPMENT AND PRODUCTION, 2008
BROWN ET AL.: "Use of Charged Membranes to Identify Soluble Protein Foulants in order to Facilitate Parvovirus Filtration", IBC'S 20TH ANTIBODY DEVELOPMENT AND PRODUCTION, 2008
BROWN, DEV. BIOL. STAND., vol. 81, 1993
BRUGGERMANN ET AL., YEAR IN IMMUNO., vol. 7, 1993, pages 33
CAPEL ET AL., IMMUNOMETHODS, vol. 4, 1994, pages 25 - 34
CARTER ET AL.: "89", PROC. NATL. ACAD. SCI. USA, 1992, pages 4285
CHOTHIA ET AL., J. MOL. BIOL., vol. 196, 1987, pages 901
CHOTHIA, C. ET AL., J. MOL. BIOL., vol. 196, 1987, pages 901 - 917
CHU; ROBINSON, CURRENT OPINION IN BIOTECHNOLOGY, vol. 12, 2001, pages 180 - 187
CLACKSON ET AL., NATURE, vol. 352, 1991, pages 624 - 628
CLYNES ET AL., PNAS USA, vol. 95, 1998, pages 652 - 656
COLE ET AL., MONOCLONAL ANTIBODIES AND CANCER THERAPY, 1985, pages 77
CURTIS ET AL., BIOTECHNOLOGY AND BIOENGINEERING, vol. 84, no. 2, 2003, pages 179 - 186
FISHWILD ET AL., NATURE BIOTECHNOLOGY, vol. 14, 1996, pages 845 - 851
GARNICK, DEV BIOL STAND. BASEL: KARGER, vol. 93, 1998, pages 21 - 29
GAZZANO-SANTORO ET AL., J. IMMUNOL. METHODS, vol. 202, 1996, pages 163
GODING: "Monoclonal Antibodies: Principals and Practice", 1986, ACADEMIC PRESS, pages: 59 - 103
GODING: "Monoclonal Antibodies: Principles and Practice", 1986, ACADEMIC PRESS, pages: 59 - 103
GRAHAM ET AL., VIROLOGY, vol. 52, 1978, pages 456 - 457
GRUBER ET AL., J. IMMUNOL., vol. 152, 1994, pages 5368
GUYER ET AL., J. IMMUNOL., vol. 117, 1976, pages 587
HAAS ET AL., J. LAB. CLIN. MED., vol. 126, 1995, pages 330 - 341
HIRASAKI ET AL., POLYMER JOURNAL, vol. 26, no. 11, 1994, pages 1244 - 1256
HOLLINGER ET AL., PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 6444 - 6448
HOOGENBOOM ET AL., J. MOL. BIOL., vol. 227, 1991, pages 381
HOOGENBOOM; WINTER, J. MOL. BIOL., vol. 227, 1991, pages 381
JAKOBOVITS ET AL., NATURE, vol. 362, 1993, pages 255 - 258
JAKOBOVITS ET AL., PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 2551
JONES ET AL., NATURE, vol. 321, 1986, pages 522 - 525
KABAT ET AL.: "Sequences of Proteins of Immunological Interest", 1991, NATIONAL INSTITUTE OF HEALTH
KABAT ET AL.: "Sequences of Proteins of Immunological Interest", 1991, PUBLIC HEALTH SERVICE, NATIONAL INSTITUTES OF HEALTH
KIM ET AL., J. IMMUNOL., vol. 24, 1994, pages 249
KOHLER ET AL., NATURE, vol. 256, 1975, pages 495
KOSTELNY ET AL., J. IMMUNOL., vol. 148, no. 5, 1992, pages 1547 - 1553
KOZBOR, J. IMMUNOL., vol. 133, 1984, pages 3001
LEVY ET AL.: "Filtration in the Biopharamaceutical Industry", 1998, MARCEL DEKKER, pages: 619 - 646
LIEBER ET AL., SCIENCE, vol. 182, 1973, pages 56 - 59
LONBERG ET AL., NATURE, vol. 368, 1994, pages 856 - 859
LONBERG; HUSZAR, INTERN. REV. IMMUNOL., vol. 13, 1995, pages 65 - 93
LUBINIECKI ET AL., DEV BIOL STAND, vol. 70, 1989, pages 187 - 191
M. DAERON, ANNU. REV. IMMUNOL., vol. 15, 1997, pages 203 - 234
MARKS ET AL., BIO/TECHNOLOGY, vol. 10, 1992, pages 779 - 783
MARKS ET AL., J. MOL. BIOL., vol. 222, 1991, pages 581
MARKS ET AL., J. MOL. BIOL., vol. 222, 1991, pages 581 - 597
MASSEY, NATURE, vol. 328, 1987, pages 457 - 458
MCCAFFERTY ET AL., NATURE, vol. 348, 1990, pages 552 - 554
MEHTA ET AL.: "Purifying Therapeutic Monoclonal Antibodies", CHEMICAL ENGINEERING PROGRESS, vol. 104, no. 5, 2008, pages S14 - S20, XP055315439
MILLSTEIN ET AL., NATURE, vol. 305, 1983, pages 537 - 539
MORRISON ET AL., PROC. NATL ACAD. SCI. USA, vol. 81, 1984, pages 6851
MORRISON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 81, 1984, pages 6851 - 6855
MORRISON, NATURE, vol. 368, 1994, pages 812 - 813
MUNSON ET AL., ANAL. BIOCHEM., vol. 107, 1980, pages 220
NEUBERGER ET AL., NATURE, vol. 312, 1984, pages 604 - 608
NEUBERGER, NATURE BIOTECHNOLOGY, vol. 14, 1996, pages 826
OMAR; KEMPF, TRANSFUSION, vol. 42, no. 8, 2002, pages 1005 - 1010
PLIICKTHUN, IMMUNOL. REVS., vol. 130, 1992, pages 151 - 188
PRESTA ET AL., J. IMMNOL., vol. 151, 1993, pages 2623
PRESTA, CURR. OP. STRUCT. BIOL., vol. 2, 1992, pages 593 - 596
PRESTA, CURR. OPIN. STRUCT. BIOL., vol. 2, 1992, pages 593 - 596
RAVETCH; KINET, ANNU. REV. IMMUNOL., vol. 9, 1991, pages 457 - 492
REICHMANN ET AL., NATURE, vol. 332, 1988, pages 323 - 329
RIECHMANN ET AL., NATURE, vol. 332, 1988, pages 323 - 327
RIECHMANN ET AL., NATURE, vol. 332, 1988, pages 323 - 329
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual", COLD SPRING HARBOR PRESS
SHALABY ET AL., J. EXP. MED., vol. 175, 1992, pages 217 - 225
SHAW ET AL., GENE, vol. 23, 1983, pages 315
SIMS ET AL., J. IMMUNOL., vol. 151, 1993, pages 2296
SKERRA ET AL., CURR. OPINION IN IMMUNOL., vol. 5, 1993, pages 256 - 262
SURESH ET AL., METHODS IN ENZYMOLOGY, vol. 121, 1986, pages 210
THOMAS, LANCET, vol. 343, 1994, pages 1583 - 1584
TRAUNECKER ET AL., EMBO J., vol. 10, 1991, pages 3655 - 3659
TUTT ET AL., J. IMMUNOL., vol. 147, 1991, pages 60
VAN REIS ET AL.: "Bioprocess membrane technology", JOURNAL OF MEMBRANE SCIENCES, vol. 297, no. 1-2, 2007, pages 16 - 50, XP022086062
VERHOEYEN ET AL., SCIENCE, vol. 239, 1988, pages 1534 - 1536
WATERHOUSE ET AL., NUC. ACIDS. RES., vol. 21, 1993, pages 2265 - 2266
ZAPATA ET AL., PROTEIN ENG., vol. 8, no. 10, 1995, pages 1057 - 1062

Also Published As

Publication number Publication date
NO2462158T3 (fr) 2018-06-09
JP2017206529A (ja) 2017-11-24
PL2462158T3 (pl) 2018-05-30
HUE038451T2 (hu) 2018-10-29
IL217740A0 (en) 2012-03-29
CA2770235A1 (fr) 2011-03-17
NZ597809A (en) 2014-05-30
KR20190018041A (ko) 2019-02-20
ZA201200626B (en) 2013-06-26
PT2462158T (pt) 2018-02-23
BR112012002574A2 (pt) 2020-11-03
MY188566A (en) 2021-12-22
KR20180035936A (ko) 2018-04-06
SG178276A1 (en) 2012-03-29
US20240083980A1 (en) 2024-03-14
AU2010292897A1 (en) 2012-02-16
MX346115B (es) 2017-03-08
IL273486B (en) 2022-04-01
AU2010292897B2 (en) 2016-01-07
US20200347118A1 (en) 2020-11-05
SI2462158T1 (en) 2018-04-30
HRP20180309T1 (hr) 2018-03-23
US10662237B2 (en) 2020-05-26
LT2462158T (lt) 2018-03-26
JP2013501075A (ja) 2013-01-10
KR20130051431A (ko) 2013-05-20
MX2012001659A (es) 2012-04-30
ES2662529T3 (es) 2018-04-06
CN102712691A (zh) 2012-10-03
IL217740B (en) 2020-04-30
RU2573894C2 (ru) 2016-01-27
CN106905413A (zh) 2017-06-30
US20110034674A1 (en) 2011-02-10
EP2462158B1 (fr) 2018-01-10
JP2023029853A (ja) 2023-03-07
KR101844859B1 (ko) 2018-05-18
RU2012108318A (ru) 2013-09-20
DK2462158T3 (en) 2018-03-05
US11225513B2 (en) 2022-01-18
IL287431A (en) 2021-12-01
IL273486A (en) 2020-05-31
JP2016106096A (ja) 2016-06-16
EP2462158A1 (fr) 2012-06-13
WO2011031397A1 (fr) 2011-03-17
US20220306726A1 (en) 2022-09-29
CA2770235C (fr) 2021-06-29
JP2020023491A (ja) 2020-02-13

Similar Documents

Publication Publication Date Title
US11225513B2 (en) Method to improve virus filtration capacity
KR100960211B1 (ko) 이온 교환 크로마토그래피에 의한 단백질 정제 방법
EP1543038B2 (fr) Purification de proteines
ES2527943T3 (es) Métodos para eliminar un contaminante usando cromatografía de membrana de intercambio iónico de desplazamiento de proteínas
KR20120106719A (ko) 변형된 단백질 a 용리를 통한 증진된 단백질 정제
TW202112799A (zh) 過載層析管柱之再生方法

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN PUBLISHED

AC Divisional application: reference to earlier application

Ref document number: 2462158

Country of ref document: EP

Kind code of ref document: P

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK SM TR

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20181018

RBV Designated contracting states (corrected)

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK SM TR

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20200703

TPAC Observations filed by third parties

Free format text: ORIGINAL CODE: EPIDOSNTIPA

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS